Dasatinib response in acute myeloid leukemia is correlated with FLT3/ITD, PTPN11 mutations and a unique gene expression signature

Sigal Tavor, Tali Shalit, Noa Chapal Ilani, Yoni Moskovitz, Nir Livnat, Yoram Groner, Haim Barr, Mark D Minden, Alexander Plotnikov, Michael W Deininger, Nathali Kaushansky, Liran I Shlush, Sigal Tavor, Tali Shalit, Noa Chapal Ilani, Yoni Moskovitz, Nir Livnat, Yoram Groner, Haim Barr, Mark D Minden, Alexander Plotnikov, Michael W Deininger, Nathali Kaushansky, Liran I Shlush

Abstract

Novel targeted therapies demonstrate improved survival in specific subgroups (defined by genetic variants) of acute myeloid leukemia (AML) patients, validating the paradigm of molecularly targeted therapy. However, identifying correlations between AML molecular attributes and effective therapies is challenging. Recent advances in high-throughput in vitro drug sensitivity screening applied to primary AML blasts were used to uncover such correlations; however, these methods cannot predict the response of leukemic stem cells (LSCs). Our study aimed to predict in vitro response to targeted therapies, based on molecular markers, with subsequent validation in LSCs. We performed ex vivo sensitivity screening to 46 drugs on 29 primary AML samples at diagnosis or relapse. Using unsupervised hierarchical clustering analysis we identified group with sensitivity to several tyrosine kinase inhibitors (TKIs), including the multi-TKI, dasatinib, and searched for correlations between dasatinib response, exome sequencing and gene expression from our dataset and from the Beat AML dataset. Unsupervised hierarchical clustering analysis of gene expression resulted in clustering of dasatinib responders and non-responders. In vitro response to dasatinib could be predicted based on gene expression (AUC=0.78). Furthermore, mutations in FLT3/ITD and PTPN11 were enriched in the dasatinib sensitive samples as opposed to mutations in TP53 which were enriched in resistant samples. Based on these results, we selected FLT3/ITD AML samples and injected them to NSG-SGM3 mice. Our results demonstrate that in a subgroup of FLT3/ITD AML (4 out of 9) dasatinib significantly inhibits LSC engraftment. In summary we show that dasatinib has an anti-leukemic effect both on bulk blasts and, more importantly, LSCs from a subset of AML patients that can be identified based on mutational and expression profiles. Our data provide a rational basis for clinical trials of dasatinib in a molecularly selected subset of AML patients.

Figures

Figure 1.
Figure 1.
In vitro drug resistance. (A) In vitro drug resistance to most drugs is acquired after relapse. An average drug response of seven couples of primary acute myeloid leukemia (AML) samples, at diagnosis (DX) and relapse (REL) is shown, comparing the half maximal inhibitory concentration (IC50) of 41 drugs. Each dot represents the response to a specific drug, calculated by the median IC50 ratio of diagnosis vs. relapse. (B) Drug sensitivity and resistance testing (DSRT) of primary AML cells. Hierarchical clustering using Pearson dissimilarity and complete linkage was performed. Data are the log2 IC50 +0.001, standardized for each compound by reducing the mean. DSRT for 41 drugs of clinical and preclinical use in AML shows two clustered groups of patients. The age and gender of the patients and the origin of the patients’ diagnostic or relapse sample, the name of the drugs, the class of the drug, karyotype and mutation status for FLT3/ITD and NPM1 are shown.
Figure 2.
Figure 2.
Transcriptome profiling of dasatinib responder samples. Gene expression analysis of whole transcriptome mRNA sequencing comparing dasatinib sensitive to non-sensitive samples of acute myeloid leukemia (AML). (A) Differentially expressed genes between "responders", and "non-responders". (B) In order to extend the tested samples we also applied the same analysis to AML patients’ samples from the Beat AML dataset.
Figure 3.
Figure 3.
Analysis of differentially expressed genes between dasatinib responders and non-responders from the INCPM and Beat AML datasets. (A) Intersection of upregulated (upper panel) and downregulated (lower panel) differentially expressed genes from the INCPM and Beat AML cohorts. (B) Pathway analysis of intersecting upregulated genes in both cohorts identified significant enrichment of genes that are co-expressed with several dasatinib targets. Dasatinib targets (CSF1R, SRC, BLK) are shown in red and the asterisk marks significant enrichment (false discovery rate<0.05).
Figure 4.
Figure 4.
Prediction model to identify dasatinib responders based on gene expression. (A, B) The k-nearest neighbor (KNN) algorithm with cosine similarity was used to predict the response to dasatinib. The greatest accuracy was achieved with 10- fold cross-validation applied to the differentially downregulated genes from the Beat AML dataset. Accordingly, a sensitivity of 0.85 and a specificity of 0.7 were achieved. (C) Validation of the KNN cosine prediction model on INCPM data.
Figure 5.
Figure 5.
Correlation between FLT3/ITD, PTPN11 and TP53 mutations in acute myeloid leukemia samples and response to dasatinib in vitro in the Beat AML study. (A-C) Median dasatinib half maximal inhibitory concentration (IC50) values in cases with mutated FLT3/ITD (A), PTPN11 (B) or TP53 (C) and wild-type (WT) samples. Differences between groups were validated by the Wilcoxon rank-sum test.
Figure 6.
Figure 6.
Dasatinib delays development of human FLT3/ITD-mutated acute myeloid leukemia in transplanted mice. SGM3 mice were treated with dasatinib for 3 weeks after transplantation of human cells from nine patients with FLT3/ITD acute myeloid leukemia (AML). The percentage of human CD45+ (hCD45) cells in engrafted murine bone marrow with/without dasatinib treatment is shown after staining for CD45, CD34, CD33, CD38, CD15, CD3 and CD19 to determine myeloid lineage cells and analyzed by fluorescence- activated cell sorting immunostaining. Wilcoxon rank sum test, *P<0.05; **P<0.005; ***P<0.0005. The mutation status of the AML samples was: 140005: FLT3/ITD+, FLT3/TKD-, NPM1+; 40094: NPM1+, FLT3/ITD+; 150809: NPM1-, FLT3/ITD+; 40034: NPM1-, FLT3/ITD+, 150279: FLT3/ITD+, FLT3/TKD-, NPM1+; 130695: NPM1+, FLT3/ITD+, FLT3/TKD-; 160436: NPM1+, FLT3/ITD+; 160406: FLT3/ITD+, NPM1+; 130607: FLT3/ITD+, NPM1+.
Graphical Abstract
Graphical Abstract

References

    1. Tuval A, Shlush LI. Evolutionary trajectory of leukemic clones and its clinical implications. Haematologica. 2019;104(5):872-880.
    1. McMahon CM, Perl AE. Gilteritinib for the treatment of relapsed and/or refractory FLT3- mutated acute myeloid leukemia. Expert Rev Clin Pharmacol. 2019;12(9):841-849.
    1. Perl AE. The role of targeted therapy in the management of patients with AML. Blood Adv. 2017;1(24):2281-2294.
    1. Stone RM, Larson RA, Dohner H. Midostaurin in FLT3-mutated acute myeloid leukemia. N Engl J Med. 2017;377(19):1903.
    1. DiNardo CD. Ivosidenib in IDH1-mutated acute myeloid leukemia. N Engl J Med. 2018;379(12):1186.
    1. Stein EM, DiNardo CD, Pollyea DA, et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood. 2017;130(6):722-731.
    1. Bisaillon R, Moison C, Thiollier C, et al. Genetic characterization of ABT-199 sensitivity in human AML. Leukemia. 2020;34(1): 63-74.
    1. Pollyea DA, Stevens BM, Jones CL, et al. Venetoclax with azacitidine disrupts energy metabolism and targets leukemia stem cells in patients with acute myeloid leukemia. Nat Med. 2018;24(12):1859-1866.
    1. Shlush LI, Mitchell A, Heisler L, et al. Tracing the origins of relapse in acute myeloid leukaemia to stem cells. Nature. 2017;547(7661):104-108.
    1. Shlush LI, Chapal-Ilani N, Adar R, et al. Cell lineage analysis of acute leukemia relapse uncovers the role of replication-rate heterogeneity and microsatellite instability. Blood. 2012;120(3):603-612.
    1. Ding L, Ley TJ, Larson DE, et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature. 2012;481(7382):506-510.
    1. Estey E, Levine RL, Lowenberg B. Current challenges in clinical development of "targeted therapies": the case of acute myeloid leukemia. Blood. 2015;125(16):2461-2466.
    1. Eriksson A, Osterroos A, Hassan S, et al. Drug screen in patient cells suggests quinacrine to be repositioned for treatment of acute myeloid leukemia. Blood Cancer J. 2015;5:e307.
    1. Snijder B, Vladimer GI, Krall N, et al. Imagebased ex-vivo drug screening for patients with aggressive haematological malignan cies: interim results from a single-arm, openlabel, pilot study. Lancet Haematol. 2017;4(12):e595-e606.
    1. Pemovska T, Kontro M, Yadav B, et al. Individualized systems medicine strategy to tailor treatments for patients with chemorefractory acute myeloid leukemia. Cancer Discov. 2013;3(12):1416-1429.
    1. Bose P, Vachhani P, Cortes JE. Treatment of Relapsed/Refractory Acute Myeloid Leukemia. Curr Treat Options Oncol. 2017;18(3):17.
    1. Tyner JW, Tognon CE, Bottomly D, et al. Functional genomic landscape of acute myeloid leukaemia. Nature. 2018;562(7728): 526-531.
    1. Gerstung M, Papaemmanuil E, Martincorena I, et al. Precision oncology for acute myeloid leukemia using a knowledge bank approach. Nat Genet. 2017;49(3):332-340.
    1. Chen EY, Tan CM, Kou Y, et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics. 2013;14:128.
    1. Kuleshov MV, Jones MR, Rouillard AD, et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016;44(W1): W90-97.
    1. Anders S, McCarthy DJ, Chen Y, et al. Count-based differential expression analysis of RNA sequencing data using R and Bioconductor. Nat Protoc. 2013;8(9):1765-1786.
    1. Megias-Vericat JE, Martinez-Cuadron D, Sanz MA, et al. Salvage regimens using conventional chemotherapy agents for relapsed/refractory adult AML patients: a systematic literature review. Ann Hematol. 2018;97(7):1115-1153.
    1. Stone RM, Mandrekar SJ, Sanford BL, et al. Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation. N Engl J Med. 2017;377(5):454-464.
    1. Fiegl M, Unterhalt M, Kern W, et al. Chemomodulation of sequential high-dose cytarabine by fludarabine in relapsed or refractory acute myeloid leukemia: a randomized trial of the AMLCG. Leukemia. 2014;28(5):1001-1007.
    1. DiNardo CD, Pratz K, Pullarkat V, et al. Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood. 2019;133(1):7-17.
    1. Papaemmanuil E, Gerstung M, Bullinger L, et al. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374(23):2209-2221.
    1. Sanchez PV, Perry RL, Sarry JE, et al. A robust xenotransplantation model for acute myeloid leukemia. Leukemia. 2009;23(11): 2109-2117.
    1. Kottaridis PD, Gale RE, Frew ME, et al. The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood. 2001;98(6):1752-1759.
    1. Yanada M, Matsuo K, Suzuki T, et al. Prognostic significance of FLT3 internal tandem duplication and tyrosine kinase domain mutations for acute myeloid leukemia: a meta-analysis. Leukemia. 2005;19(8):1345-1349.
    1. Thiede C, Steudel C, Mohr B, et al. Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis. Blood. 2002;99(12):4326-4335.
    1. Kayser S, Schlenk RF, Londono MC, et al. Insertion of FLT3 internal tandem duplication in the tyrosine kinase domain-1 is associated with resistance to chemotherapy and inferior outcome. Blood. 2009;114(12):2386-2392.
    1. Sasaki K, Kantarjian HM, Kadia T, et al. Sorafenib plus intensive chemotherapy improves survival in patients with newly diagnosed, FLT3-internal tandem duplication mutation-positive acute myeloid leukemia. Cancer. 2019;125(21):3755-3766.
    1. Uy GL, Mandrekar SJ, Laumann K, et al. A phase 2 study incorporating sorafenib into the chemotherapy for older adults with FLT3-mutated acute myeloid leukemia: CALGB 11001. Blood Adv. 2017;1(5):331-340.
    1. Rollig C, Serve H, Huttmann A, et al. Addition of sorafenib versus placebo to standard therapy in patients aged 60 years or younger with newly diagnosed acute myeloid leukaemia (SORAML): a multicentre, phase 2, randomised controlled trial. Lancet Oncol. 2015;16(16):1691-1699.
    1. Weisberg E, Liu Q, Nelson E, et al. Using combination therapy to override stromalmediated chemoresistance in mutant FLT3- positive AML: synergism between FLT3 inhibitors, dasatinib/multi-targeted inhibitors and JAK inhibitors. Leukemia. 2012;26(10):2233-2244.
    1. Saito Y, Kitamura H, Hijikata A, et al. Identification of therapeutic targets for quiescent, chemotherapy-resistant human leukemia stem cells. Sci Transl Med. 2010;2(17):17ra9.
    1. Dos Santos C, McDonald T, Ho YW, et al. The Src and c-Kit kinase inhibitor dasatinib enhances p53-mediated targeting of human acute myeloid leukemia stem cells by chemotherapeutic agents. Blood. 2013;122(11):1900-1913.
    1. Kazi JU, Ronnstrand L. The role of SRC family kinases in FLT3 signaling. Int J Biochem Cell Biol. 2019;107:32-37.
    1. Li L, Modi H, McDonald T, et al. A critical role for SHP2 in STAT5 activation and growth factor-mediated proliferation, survival, and differentiation of human CD34+ cells. Blood. 2011;118(6):1504-1515.
    1. Schubbert S, Lieuw K, Rowe SL, et al. Functional analysis of leukemia-associated PTPN11 mutations in primary hematopoietic cells. Blood. 2005;106(1):311-317.
    1. Jenkins C, Luty SB, Maxson JE, et al. Synthetic lethality of TNK2 inhibition in PTPN11-mutant leukemia. Sci Signal. 2018;11(539):eaao5617.

Source: PubMed

3
Se inscrever